Вы находитесь на странице: 1из 10

The Pathophysiology and Treatment

of Hereditary Tyrosinemia Type 1

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
Markus Grompe, M.D.1

ABSTRACT

The topic of this review is hepatorenal tyrosinemia (hereditary tyrosinemia type


1 [HT1], or fumarylacetoacetate hydrolase deficiency; OMIM# 276700). HT1 is the
most serious and common of the genetic defects in tyrosine degradation. In addition, this
disorder has importance as a model of spontaneous self-correction of liver disease, as a
model of liver repopulation by transplanted cells and gene therapy, and as a genetic cause
of hepatocarcinoma. However, other forms of hypertyrosinemia exist; hence, the differen-
tial diagnosis also will be described briefly. Recent years have seen much progress in our
understanding of the molecular basis, the pathophysiology, and especially the treatment
of HT1. The current intervention with 2-(2-nitro-4-trifluoro-methylbenzyol)-1,3 cyclo-
hexanedione (NTBC) therapy has improved the outcome of this once devastating disor-
der. The successful repopulation of the HT1 liver with transplanted cells and positive re-
sults in the use of gene therapy in animal models may someday lead to therapy in humans
that will obviate the need for life-long dietary and pharmacological therapy.

KEYWORDS: Tyrosinemia, succinylacetone, hepatocarcinoma therapy

Objectives: Upon completion of this article the reader should be able to (1) describe the differential diagnosis of hypertyrosinemia,
(2) list the therapeutic approaches in hepatorenal tyrosinemia, and (3) describe the pathophysiology of hepatorenal tyrosinemia.
Accreditation: Tufts University School of Medicine is accredited by the Accreditation Council for Continuing Medical Education to
provide continuing medical education for physicians. TUSM takes full responsibility for the content, quality, and scientific integrity of
this continuing education activity.
Credit: Tufts University School of Medicine designates this education activity for a maximum of 1.0 hour credit toward the AMA
Physicians Recognition Award in category one. Each physician should claim only those hours that he/she actually spent in the edu-
cational activity.

I mpaired degradation of the aromatic amino types, the hepatocyte and renal proximal tubules, ex-
acid tyrosine is a feature of several acquired and genetic press the complete pathway and contain sufficient
liver disorders. In humans, hypertyrosinemia is defined quantities of all the enzymes required for tyrosine ca-
by elevated blood levels of >200 M.1 In mammals, ty- tabolism. Autosomal-recessive enzyme deficiency dis-
rosine is both ketogenic and glucogenic. Its degradation eases have been described for four of the five degrada-
is catalyzed by a series of five enzymatic reactions tion steps, but not all of these result in elevated blood
(Fig. 1) yielding acetoacetate (ketogenic) and the Krebs tyrosine levels. Only one of these disorders, hereditary
cycle intermediate fumarate (glucogenic). Only two cell tyrosinemia type 1 (HT1) causes liver disease and

Seminars in Liver Disease, volume 21, number 4, 2001. Address for correspondence and reprint requests: Markus Grompe, M.D., Department of
Molecular and Medical Genetics, L103, Oregon Health Sciences University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97201. E-mail:
grompem@ohsu.edu. 1Department of Molecular and Medical Genetics, Oregon Health Sciences University, Portland, Oregon. Copyright
2001 by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA. Tel: +1(212) 584-4662. 0272-8087,p;2001,21,04,
563,572,ftx,en;sld00145x.
563
564 SEMINARS IN LIVER DISEASE/VOLUME 21, NUMBER 4 2001

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
Figure 1 The mammalian tyrosine catabolic pathway. TAT, tyrosine amino transferase; HPD, 4-hydroxyphenylpyruvate dioxygenase;
HGD, homogentisic acid dioxygenase; MAI, maleylacetoacetate isomerase; FAH, fumarylacetoacetate hydrolase; FAR, fumarylace-
toacetate reductase; HT1, hereditary tyrosinemia type 1. The enzymatic reactions deficient in this disease are indicated by the hori-
zontal bar. NTBC, 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione.

therefore will be the main focus of this review. The DIAGNOSIS AND
reader is referred elsewhere1 for details on the other dis- DIFFERENTIAL DIAGNOSIS OF
orders of tyrosine degradation. The most common form HYPERTYROSINEMIA
of tyrosinemia is transient tyrosinemia of the newborn, To detect elevated blood tyrosine levels, quantitative
which is particularly common in premature infants and measurement of plasma amino acids is required. This
results from delayed maturation of the tyrosine metabo- test is usually performed to rule out metabolic liver dis-
lizing enzymes.2 Elevated blood tyrosine is also found ease or to evaluate possible metabolic etiologies in a
in scurvy and many forms of both acute and chronic ac- child with developmental delay. In some situations, ele-
quired liver disease. vated blood tyrosines are discovered because of elevated
HEREDITARY TYROSINEMIA/GROMPE 565

Table 1 Etiology of Elevated Blood Tyrosine Levels ACQUIRED TYROSINEMIA


1. Hepatocellular dysfunction
Increased blood tyrosine levels most commonly have
2. Transient tyrosinemia of the newborn
nongenetic origins. Elevated tyrosine per se does not
3. Genetic enzyme deficiencies in tyrosine degradation
cause any disease when the blood levels remain below
Hepatorenal tyrosinemia (HT1)
500 M, and hypertyrosinemia is therefore more of di-
Oculocutaneous tyrosinemia (HT2)
agnostic interest than of therapeutic concern.
4-OH-phenylpyruvate dioxygenase deficiency (HT3)
The most common noninherited cause of in-

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
4. Miscellaneous
creased blood tyrosine levels is transient tyrosinemia of
Scurvy
the newborn.1,2 This condition is due to immaturity
Hyperthyroidism
of the liver and the enzymes involved in tyrosine degra-
Postprandial sample
dation. Historically, transient tyrosinemia was a com-
NTBC therapy
mon finding, but it has become much more rare with
modern neonatal management. When present it re-
sponds rapidly to pharmacological doses of vitamin C.
Hepatocellular dysfunction of any cause can re-
tyrosine in a urine metabolic screen (renal Fanconi syn- sult in elevated blood tyrosine and excretion of in-
drome). The genetic and acquired disorders that can creased amounts of tyrosine metabolites in urine. The
lead to tyrosinemia are listed in Table 1. The main clin- tyrosine levels are usually under 500 M, similar to
ical manifestations of hereditary disorders in tyrosine what is seen in hereditary tyrosinemia type 1. For this
degradation are listed in Table 2. The presence or ab- reason, measurement of AFP levels and urinary suc-
sence of liver disease is the most important considera- cinylacetone are mandatory in this setting.
tion for both diagnosis and treatment. If liver disease is
present, additional diagnostic tests are urgently needed
to determine whether the patient has hepatorenal ty- TYROSINEMIA TYPE 1
rosinemia (fumarylacetoacetate hydrolase deficiency is
the equivalent of tyrosinemia type 1). These tests in- Clinical Manifestations
clude measurement of plasma levels of !-fetoprotein The natural history of HT1 is characterized by severe
(AFP) and the analysis of urine organic acids to check liver disease, which frequently results in death. Symp-
for the presence succinylacetone, a hallmark metabolite toms can present early in infancy with an acute rapid
of this disease. course, or they may progress more chronically.3 Interest-

Table 2 Clinical Manifestations of Enzyme Deficiencies in Tyrosine Catabolism


Elevated Renal
Defective Blood Corneal Hyper- Mental Liver Fanconi
Enzyme Disease Names Tyrosine Ulcers keratosis Retardation Arthritis Failure Syndrome

Tyrosine Tyrosinemia type II +++ +++ ++ +/"


aminotransferase Oculocutaneous
tyrosinemia
Richner-Hanhardt
syndrome
4-OH- Tyrosinemia type III ++ +/" +/" +/"
phenylpyruvate
dioxygenase
Homogentisate Alkaptonuria +++
dioxygenase
Maleylacetoacetate No human patients ? ? ? ? ? ? ?
isomerase described
Fumarylacetoacetate Tyrosinemia type I ++ +++ ++
hydrolase Hepatorenal
tyrosinemia
, symptom absent; +/", symptom present in some patients; +, ++, +++, symptom always present, ranging from mild (+) to moderate
(++) to severe (+++).
566 SEMINARS IN LIVER DISEASE/VOLUME 21, NUMBER 4 2001

ingly, the acute and chronic forms of the disease can tions.11 A simple polymerase chain reaction (PCR)-
occur within the same sibship. Most patients present based method for the detection of IVS12 + 5 g a has
with failure to thrive and hepatomegaly. The liver disease been developed and could be applied to population
is progressive, causing micro- and macronodular cirrho- screening. Another splice mutation, IVS6-1 g t, is
sis.4 Icterus, ascites, and hemorrhage often ensue. Pa- also fairly common and has been found in several popu-
tients also display renal tubular acidosis of the Fanconi lations. The Finnish HT1 founder mutation (W262X)
type, and typical radiographic changes of rickets are often is a missense change.10 An up-to-date listing of known

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
present. Mental retardation is not a feature. Surviving pa- FAH mutations can be found at the Human Gene Mu-
tients have a high risk for developing hepatocarcinoma.4 tation Database Web site (archive.uwcm.ac.uk/uwcm/
Porphyria-like neurologic crises are common in mg/search/119901.html).
poorly controlled HT1 and can be a major cause of Prenatal diagnosis for HT1 can be made using a
morbidity and mortality.5 The attacks can present as variety of methods. The presence of succinylacetone in
pain, obtundation, and progressive paralysis, similar to amniotic fluid is a fairly reliable indicator of the disease,
classic acute intermittent porphyria. but false-negative results have been reported.12,13 The
FAH enzyme also can also be measured directly in cul-
tured amniocytes.14 Today, accurate DNA-based diag-
Laboratory Diagnosis nosis is possible using both linkage analysis or direct
Biochemical alterations include elevated plasma con- mutation detection.15 The human FAH gene has several
centrations of tyrosine and methionine, as well as the intragenic polymorphic markers, making linkage analy-
excretion of tyrosyl compounds in the urine. The pres- sis feasible in most cases.1619
ence of succinylacetone in urine is diagnostic; therefore,
urine organic acid analysis is the single most important
diagnostic test for this disease. Highly elevated concen- Newborn Screening
trations of AFP are seen, even before the elevation in Newborn screening for HT1 is performed in some re-
tyrosine. Hypoglycemia may occur, and coagulation de- gions, particularly the Canadian province of Qubec,
fects are common. Plasma transaminase levels (ALT where the disease is common.20,21 With the availability
and AST) may be only mildly elevated and are in dis- of effective therapy, early identification of affected chil-
proportion to the degree of coagulopathy.6 dren can prevent the evolution of severe liver disease
Fumarylacetoacetate hydrolase (FAH) enzyme and greatly improve outcome.
activity can be measured in cultured skin fibroblasts or
liver specimens to confirm the diagnosis.
Treatment
The classic dietary treatment of tyrosinemia type 1
Genetics and Prenatal Diagnosis consisted of dietary restriction of phenylalanine and ty-
Hereditary tyrosinemia type 1 is a common inborn error rosine,22 but this regimen did not prevent progression of
of metabolism in some ethnic groups, most notably in the disease. Liver transplantation has been successfully
the population of the Lac-St. Jean region of Qubec, used in many patients.2325 The beneficial effects of liver
where the frequency of heterozygotes has been esti- transplantation strongly suggest that expression of a
mated to be 7%.7 The human FAH cDNA has been normal cDNA in hepatocytes of patients would allevi-
cloned8 and mapped to human chromosome 15q,8 and ate the defect; therefore, HT1 is a candidate disorder for
the gene structure has been elucidated.9 The human gene therapy directed at the liver.26
FAH cDNA consists of 419 amino acids encoded by a
1,260-bp mRNA. The gene is approximately 35 kbp in
size and consists of 14 exons. DNA-based prenatal di- NTBC
agnosis and carrier detection are available, and the com- The history of the discovery of 2-(2-nitro-4-trifluoro-
mon mutations responsible for the ethnic clustering of methylbenzyol)-1,3 cyclohexanedione (NTBC) as the
HT1 in French Canada and Scandinavia have been treatment for HT1 is an interesting saga. The class of
identified.10,11 compounds to which NTBC belongs was developed in
The French Canadian HT1 founder mutation is the 1980s as a bleaching herbicide by the British phar-
a single base change in intron 12 (IVS12 + 5 g a), maceutical company ICN-Zeneca. During small animal
which profoundly alters mRNA splicing and results in toxicology studies, it was noted that treated rats devel-
the absence of hepatic enzyme activity. In Qubec this oped corneal ulcerations, a hallmark of elevated blood
allele is found on more than 95% of HT1 chromo- tyrosine in both rats and humans. It was then found
somes, and this mutation is also the most common one that NTBC and similar compounds are potent in-
worldwide, accounting for about one third of all muta- hibitors of 4-OH phenylpyruvate dioxygenase (HPD),
HEREDITARY TYROSINEMIA/GROMPE 567

the second step in tyrosine catabolism (Fig. 1). Inhibi- Somatic Mosaicism and Liver Repopulation
tion of this enzyme is also the mechanism by which in Hereditary Tyrosinemia
benzoyl-cyclo-hexane-1,3-diones act as herbicides be- In 1993 an unusual observation was made in human
cause plants need homogentisic acid as precursors for patients with HT1: patient livers were removed at the
chlorophyll biosynthesis.27 A weaker analogue is cur- time of liver transplantation and multiple large and
rently marketed as an herbicide under the brand name small nodules were found as is typical in HT1. An im-
Mikado (Bayer, Leverkusen, Germany). munohistological analysis with FAH antibody was

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
In 1992, Lindstedt, Holme, and co-workers re- performed, and, surprisingly, the livers represented a
alized NTBCs potential for the treatment of human mosaic of antibody positive and negative hepatic tis-
tyrosinemia and used it successfully in five patients.28 sue.33 FAH enzyme activity was present in the cross-
The principal underlying this treatment is to reduce reacting material-positive nodules, and molecular
metabolic flow through the tyrosine catabolic pathway analysis has revealed they were generated by somatic
and to prevent the formation of the toxic compounds reversion of the disease causing inherited mutation34
maleylacetoacetate (MAA) and fumarylacetoacetate followed by clonal selection of reverted hepatocytes.
(FAA) (Fig. 1). In the most recently published study, This observation demonstrated the strong selective
over 300 patients worldwide have received this treat- growth advantage of spontaneously reverted FAH-
ment, and in most patients (>95%) liver functions im- positive hepatocytes in this disease. Human HT1 thus
proved rapidly. This represents a significant improve- provided the first example of spontaneous gene ther-
ment over historical controls, so NTBC currently is apy in the liver. Interestingly, mutation reversion has
the clear treatment of choice for HT1. Kidney func- been observed not only in compound heterozygotes, in
tion is also significantly improved by the drug, and whom intragenic mitotic recombination could result in
renal Fanconi syndrome is avoided. A large number of restoration of enzyme activity. Reversions also have
patients has now been treated for more than 5 years, been found in patients homozygous for the same mu-
and significant side effects of NTBC have not been tant allele on both chromosomes.35 This phenomenon
reported. At this writing, NTBC is still an investiga- requires precise removal of the single base change mu-
tive drug, but is available through research protocols in tation responsible for the disease.
the United States (crscott@u.washington.edu), Canada In most patients the amount of reverted tissue is
(mitchell@justine.umontreal.ca), and Europe (elisa- too low to result in a cure of the disease, but it seems
beth.holme@ clinchem.gu.se). NTBC is given twice likely that some of the intrafamilial variability in the clin-
daily orally at a typical starting dose of 1 mg/kg/day. ical course of HT1 may be due to somatic mosaicism.35
Because NTBC increases blood tyrosine levels, pa- Patients with a significant mass of reverted hepatocytes
tients also need to be kept on a protein-restricted diet may have a milder form of the disease. This hypothesis
that is low in phenylalanine and tyrosine. Effective has not yet been directly confirmed in humans, but ani-
management and adjustment of the dose requires fre- mal experiments indicate that partial reconstitution by
quent metabolic monitoring, including the measure- FAH-positive hepatocytes can be therapeutic.36
ment of plasma amino acids, blood and urinary suc-
cinylacetone, liver function tests, and plasma AFP. The
latter is particularly important because a small per- Biochemistry of FAH
centage of late-treated patients developed hepatocellu- The mammalian tyrosine catabolic pathway was worked
lar carcinoma despite NTBC treatment.29 Blood tyro- out in the early 1950s by Edwards and Knox.3739 The
sine levels should be kept under 500 M. Routine FAH enzyme assay first described then is still used
ophthalmologic examination and hepatic imaging are today. FAA is considered the natural substrate of FAH,
also recommended. Despite the clear benefits of but the enzyme also uses succinylacetoacetate (SAA) as
NTBC therapy, there are concerns about its effects a substrate (Fig. 1), and both FAA and SAA accumu-
when used long term. First, some patients have devel- late in FAH deficiency. The mechanism of the reduc-
oped hepatic cancer while on therapy. Second, all tion of FAA to SAA has not been established, but it
known patients with inherited HPD deficiency have is likely catalyzed by a yet uncharacterized enzyme,
neurologic problems.30,31 Mental retardation is also FAA-reductase.
found in 50% of tyrosinemia type II patients.32 Thus, Human wild-type FAH has a Km for FAA of
long-term follow-up studies will be required before !3.5 M. It is cytosolic and acts as a homodimer with a
the safety of this approach is fully established. molecular weight of approximately 80 kDa. Recently,
For those patients who do not respond to NTBC the crystal structure of FAH has been determined.40
therapy or have evidence of hepatic malignancy, ortho- The detailed topography of the active site was con-
topic liver transplantation remains the treatment of firmed by the development of an FAH inhibitor and
choice. cocrystallization of this molecule with the enzyme.41
568 SEMINARS IN LIVER DISEASE/VOLUME 21, NUMBER 4 2001

Pathophysiology cells display a striking disregulation of gene expression.


The discussion of the pathophysiology of HT1 will be This phenomenon was intensely studied in the lethal al-
grouped by the main findings of the disorder. bino mouse, an animal model of FAH deficiency. In
these mice, many hepatic enzymes and proteins are ex-
pressed abnormally or not at all. Interestingly, all tran-
ELEVATED BLOOD TYROSINE
scripts regulated by glucocorticoids via cyclic AMP are
In 1977 it was discovered that HT1 is caused by a defi-
severely downregulated. These include TAT,52 glucose-6
ciency of FAH, the last enzyme in tyrosine catabo-

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
phosphatase (G-6P),53 and phosphoenolpyruvate car-
lism.42 This was surprising because FAH is five steps re-
boxykinase (PEPCK).54 Other affected genes are gluta-
moved from the initial metabolism of tyrosine, and
mine synthetase (GS),53 aldolase B, and albumin.55,56 As
enzyme deficiency would not be expected to result in el-
a result, many metabolic processes are negatively af-
evated blood tyrosine. Furthermore, deficiency of ho-
fected, including gluconeogenesis, ammonia detoxifica-
mogentisic acid dioxygenase, the third enzyme of the
tion, and synthesis of secreted proteins. Other liver
degradation cascade, is not associated with hyperty-
mRNAs are increased in HT1 hepatocytes, and these
rosinemia.43 Indeed, the elevated blood tyrosine levels
include genes inducible by DNA or oxidative damage
in HT1 are due to a secondary inhibition of proximal
such as CHOP and NMO-1.57,58 The detailed mecha-
steps in tyrosine degradation and not the lack of FAH
nism by which FAH deficiency causes misexpression of
itself. In FAH mutant mice, the mRNA for tyrosine
genes is not yet understood. However, FAA acts via the
amino transferase (TAT), the rate-limiting enzyme in
electrophile response element (EPRE) DNA motif59
tyrosine degradation, is absent. The activity of HPD is
and induces phase II dioxin-inducible (Ah) genes.60
also decreased in human HT1 liver.
Thus, the perturbed gene expression can be viewed as a
Importantly, tyrosine per se is not toxic to the
protective response against severe oxidative stress.
liver or kidney, but rather causes only dermatological,
ophthalmologic, and possibly neurodevelopmental
problems.1 Patients with tyrosinemia type 2 (TAT defi- RENAL TUBULAR INJURY
ciency, Richner-Hanhardt syndrome) and type 3 (PPD Fumarylacetoacetate hydrolase deficiency can cause di-
deficiency) have highly elevated blood tyrosine levels rect injury in renal proximal tubular cells, and FAA has
but do not manifest liver disease or renal tubular induced apoptosis acutely, just as in hepatocytes.50
dysfunction.4447 However, some of the kidney phenotype is mediated by
circulating succinylacetone. This compound can induce
renal Fanconi syndrome by itself when injected into
LIVER INJURY
normal rats.61,62 Because SA is responsible for this phe-
Fumarylacetoacetate, the compound that accumulates
notype, the normalization of SA blood levels by liver
in FAH deficiency, is a potent alkylator, causing oxida-
transplantation also abrogates the renal tubular acidosis.
tive damage to the cells in which it is generated by re-
Thus, the renal pathology in HT1 appears to be largely
acting with glutathione and sulfhydryl groups of pro-
noncell autonomous.
teins. Importantly, FAA acts cell autonomously, directly
damaging only the hepatocytes and renal proximal
tubules in which it is produced and not adjacent cells.48 HEPATOCARCINOMA
Because of its rapid reactivity, FAA itself is not found in The evolution of hepatocarcinoma in human HT1 pa-
body fluids of HT1 patients. SAA and succinylacetone tients has not been studied in detail to determine
(SA) derived from reduction of FAA are the principal whether it differs from hepatocarcinoma of other
metabolites of FAA (Fig. 1). These compounds are causes. However, it is known that FAA is strongly mu-
found systemically, and testing for them diagnostically tagenic. The mutagenicity of FAA has been docu-
is routine. mented both in vitro63 and in vivo.64 The mutation
The accumulation of FAA within the hepatocyte spectrum induced by FAA in vivo in mouse hepatocytes
results in one of two outcomes: (a) apoptotic cell death has been studied in some detail. Surprisingly FAA in-
or (b) a profound perturbation of gene expression. Re- duces not only point mutations (!20%), but small in-
cent studies have shown that acute accumulation of sertions and deletions (!30%) and large genomic re-
FAA triggers apoptosis in both hepatocytes and renal arrangements (!50%).64 The precise mechanism by
tubular cells.49,50 Apoptosis is mediated by caspases which FAA causes mutations is not known. It may react
1 and 3 and is associated with cytochrome c release.51 and damage (alkylate) DNA directly, in which case
Glutathione is protective, and its intracellular level point mutations would be the most expected result. Al-
can clearly modulate the threshold for FAA-induced ternatively, FAA may act indirectly by damaging cellular
apoptosis.51 proteins involved in maintaining genomic stability. This
Hepatocytes can survive with FAH deficiency, if mechanism would provide a better explanation for the
the substrate accumulation is more gradual, but mutant mutational spectrum seen in vivo.
HEREDITARY TYROSINEMIA/GROMPE 569

NEUROLOGICAL CRISIS led to the recent discovery of extrahepatic liver stem


Several remarkable secondary biochemical alterations cells in the pancreas and bone marrow.77,78
have been described in HT1. The most prominent is the Therapeutic liver repopulation has been the sub-
inhibitory effect of SA on !-aminolevulinic acid dehy- ject of recent reviews, to which the reader is referred for
dratase, the first step of heme biosynthesis.65 This leads further details.79,80
to neurologic crisis, as seen in acute intermittent por-
phyria.5 NTBC treatment blocks the accumulation of FAH-DEFICIENT FUNGUS
SA and therefore also prevents neurologic crises.

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
A strain of FAH-deficient Aspergillus nidulans has been
instrumental in the study of tyrosine catabolism81 and
therefore will be mentioned here. Two of the genes in
Model Systems the pathway (HGD and MAI) were first cloned in this
The tyrosine catabolic pathway is conserved in many organism, and only then were then the human genes
eukaryotes, including the fungus Aspergillus nidulans, found.82,83 The molecular basis of alkaptonuria, the first
which has emerged as a important model system. Mod- human recessive disease described,84 was discovered
els of FAH deficiency have been engineered in the based on experiments performed in this system.85
mouse and in aspergillus.

THE C14CoS LETHAL ALBINO MUTANT MOUSE ABBREVIATIONS


The c14CoS albino mouse is one of a series of mutant AFP !-fetoprotein
mice bearing large overlapping x-rayinduced deletions EPRE electrophile response element
of chromosome 7.53,66 All albino deletions are estimated FAA fumarylacetoacetate
to be several megabases in size,67 but show substantial FAH fumarylacetoacetate hydrolase
overlap. Mice homozygous for the deletions c65kb, c112Kb, G-6P glucose-6 phosphatase
c3H, and c14CoS die within a few hours after birth and GS glutamine synthetase
display a striking biochemical phenotype. Profound ab- HT1 hereditary tyrosinemia type 1
normalities are found in both liver and kidney in these NTBC 2-(2-nitro-4-trifluoro-methylbenzyol)-1,3
mice, and because of this downregulation of multiple cyclohexanedione
liver enzymes in the homozygous deletion, it was hy- PEPCK phosphoenolpyruvate carboxykinase
pothesized that a regulatory liver gene (hepatocyte- PPD phenylpyruvate dioxygenase
specific developmental regulation locus, or hsdr-1) was SA succinylacetone
localized within the region.56 Detailed mapping of the SAA succinylacetoacetate
deletion intervals revealed that the hsdr-1 gene was TAT tyrosine amino transferase
identical to FAH.48 This was confirmed by the fact that
FAH knockout mice had very similar hepatic pheno-
REFERENCES
types to c14CoS albino mice. Hence, the profound disreg-
ulation of hepatic gene expression in this model can be 1. Mitchell GA, Grompe M, Lambert M, et al. Hypertyrosine-
ascribed completely to the metabolic effects of FAA mia. In: Scriver CR, Beaudet AL, Sly W, Valle D, eds. The
accumulation. Metabolic and Molecular Basis of Inherited Disease, 9th ed.
New York: McGraw-Hill, 2001:17771805
2. Levine SZ, Marples E, Gordon HH. A defect in the metabo-
lism of aromatic amino acids in premature infants: The role of
FAH#exon 5 Knockout Mice vitamin C. Science 1939;90:620
Another model of FAH deficiency in the mouse was 3. Kvittingen E. Hereditary tyrosinemia type Ian overview.
generated by targeted disruption of exon 5 of the gene Scand J Clin Lab Invest 1986;46:2734
4. Russo P, ORegan S. Visceral pathology of hereditary tyrosine-
in embryonic stem cells.68 Homozygotes had a pheno-
mia type I. Am J Hum Genet 1990;47:317324
type identical to lethal albino mice and died in the new- 5. Mitchell G, Larochelle J, Lambert M, et al. Neurologic crises
born period. Fortunately, NTBC also worked in mice in hereditary tyrosinemia. N Engl J Med 1990;322:432437
and can be used to rescue FAH mutant mice.69 NTBC- 6. Croffie JM, Gupta SK, Chong SK, et al. Tyrosinemia type 1
treated FAH#exon5 animals can be maintained for more should be suspected in infants with severe coagulopathy even
than 1 year without liver disease and are fertile.70 This in the absence of other signs of liver failure. Pediatrics 1999;
strain of mice has become one of the main models for 103:675678
7. De Braekeleer M, Larochelle J. Genetic epidemiology of he-
liver repopulation by genetically modified hepato-
reditary tyrosinemia in Quebec and in Saguenay-Lac-St-Jean.
cytes36,7173 or cell transplantation.7476 FAH-positive Am J Hum Genet 1990;47:302307
hepatocytes have a strong selective advantage in the set- 8. Phaneuf D, Labelle Y, Brub D, et al. Cloning and expression
ting of a tyrosinemic liver similar to that seen in human of the cDNA encoding human fumarylacetoacetae hydrolase,
patients.33 The strong selection in this model also has the enzyme deficient in hereditary tyrosinemia: assignment of
570 SEMINARS IN LIVER DISEASE/VOLUME 21, NUMBER 4 2001

the gene to chromosome 15. Am J Hum Genet 1991;48: (NTBC), its toxicology and development as a drug. J Inherit
525535 Metab Dis 1998;21:498506
9. Labelle Y, Phaneuf D, Leclerc B, et al. Characterization of 28. Lindstedt S, Holme E, Lock EA, et al. Treatment of heredi-
the human fumarylacetoacetate hydrolase gene and identifi- tary tyrosinaemia type I by inhibition of 4-hydroxyphenyl-
cation of a missense mutation abolishing enzymatic activity. pyruvate dioxygenase. Lancet 1992;340:813817
Hum Mol Genet 1993;2:941946 29. Holme E, Lindstedt S. Tyrosinaemia type I and NTBC (2-
10. St-Louis M, Leclerc B, Laine J, et al. Identification of a stop (2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione). J
mutation in five Finnish patients suffering from hereditary Inherit Metab Dis 1998;21:507517

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
tyrosinemia type I. Hum Mol Genet 1994;3:6972 30. Origuchi Y, Endo F, Kitano A, et al. Sural nerve lesions in a
11. Grompe M, St-Louis M, Demers SI, et al. A single mutation case of hypertyrosinemia. Brain Dev 1982;4:463
of the fumarylacetoacetate hydrolase gene in French Canadi- 31. Giardini O, Cantani A, Kennaway NG, et al. Chronic ty-
ans with hereditary tyrosinemia type I. N Engl J Med 1994; rosinemia associated with 4-hydroxyphenylpyruvate dioxyge-
331:353357 nase deficiency with acute intermittent ataxia and without
12. Jakobs C, Stellaard F, Kvittingen EA, et al. First-trimester visceral and bone involvement. Pediatr Res 1983;17:2529
prenatal diagnosis of tyrosinemia type I by amniotic fluid suc- 32. Fois A, Borgogni P, Cioni M, et al. Presentation of the data of
cinylacetone determination. Prenat Diagn 1990;10:133134 the Italian registry for oculocutaneous tyrosinaemia. J Inherit
13. Grenier A, Cederbaum S, Laberge C, et al. A case of tyrosi- Metab Dis 1986;9:262
naemia type I with normal level of succinylacetone in the am- 33. Kvittingen EA, Rootwelt H, Brandtzaeg P, et al. Hereditary
niotic fluid. Prenat Diagn 1996;16:239242 tyrosinemia type I. Self-induced correction of the fumary-
14. Kvittingen EA, Steinmann B, Gitzelmann R, et al. Prenatal lacetoacetase defect. J Clin Invest 1993;91:18161821
diagnosis of hereditary tyrosinemia by determination of fu- 34. Kvittingen EA, Rootwelt H, Berger R, et al. Self-induced
marylacetoacetase in cultured amniotic fluid cells. Pediatr Res correction of the genetic defect in tyrosinemia type I. J Clin
1985;19:334337 Invest 1994;94:16571661
15. Demers SI, Phaneuf D, Tanguay RM. Hereditary tyrosinemia 35. Poudrier J, Lettre F, Scriver CR, et al. Different clinical forms
type I: strong association with haplotype 6 in French Canadi- of hereditary tyrosinemia (type I) in patients with identical
ans permits simple carrier detection and prenatal diagnosis. genotypes. Mol Genet Metab 1998;64:119125
Am J Hum Genet 1994;55:327333 36. Overturf K, Al-Dhalimy M, Tanguay R, et al. Hepatocytes
16. Rootwelt H, Kvittingen EA, Hoie K, et al. The human fu- corrected by gene therapy are selected in vivo in a murine
marylacetoacetase gene: characterisation of restriction frag- model of hereditary tyrosinaemia type I. Nat Genet 1996;12:
ment length polymorphisms and identification of haplotypes 266273
in tyrosinemia type 1 and pseudodeficiency. Hum Genet 37. Knox WE, Edwards SW. Enzymes involved in conversion of
1992;89:229233 tyrosine to acetoacetate. Methods Enzymol 1955;2:287300
17. Demers SI, Phaneuf D, Tanguay RM. TaqI RFLP for the 38. Edwards SW, Knox WE. Homogentisate metabolism: the iso-
human fumarylacetoacetate hydrolase (FAH) gene. Nucleic merization of maleylacetoacetate by an enzyme which requires
Acids Res 1991;19:1352 glutathione. J Biol Chem 1956;220:79
18. Demers SI, Tanguay RM. MspI RFLP in the human fumary- 39. Knox WE, Edwards SW. Homogentisate oxidase of liver. J
lacetoacetate hydrolase (FAH) gene. Nucleic Acids Res 1991; Biol Chem 1955;216:479487
19:6971 40. Timm DE, Mueller HA, Bhanumoorthy P, et al. Crystal
19. Demers SI, Tanguay RM. Bg1II RFLP for the human fu- structure and mechanism of a carbon-carbon bond hydrolase.
marylacetoacetate hydrolase (FAH) gene. Nucleic Acids Res Structure Fold Des 1999;7:10231033
1991;19:1965 41. Bateman RL, Bhanumoorthy P, Witte JF, et al. Mechanistic
20. Hutchesson AC, Hall SK, Preece MA, Green A. Screening inferences from the crystal structure of fumarylacetoacetate
for tyrosinaemia type I. Arch Dis Child Fetal Neonatal Ed hydrolase with a bound phosphorus-based inhibitor. J Biol
1996;74:F191F194 Chem 2001;276:1528415291
21. Belanger M, Saint HB, Belanger L. A system of early screen- 42. Lindblad B, Lindstedt S, Steen G. On the enzymic defects in
ing for inborn errors of metabolism: methods and results for hereditary tyrosinemia. Proc Natl Acad Sci USA 1977;74:
hereditary tyrosinemia. Union Med Can 1973;102:294302 46414645
22. Bain MD, Purkiss P, Jones M, et al. Dietary treatment elimi- 43. La Du BN, Zannoni VG, Laster L, et al. The nature of the
nates succinylacetone from the urine of a patient with ty- defect in tyrosine metabolism in alkaptonuria. J Biol Chem
rosinemia type I. Eur J Pediatr 1990;149:637639 1958;230:251260
23. Murcia FJ, Vazquez J, Gamez M, et al. Liver transplantation 44. Rehak A, Selim MM, Yadav G. Richner-Hanhart syndrome
in type I tyrosinemia. Transplant Proc 1995;27:23012302 (tyrosinaemia-II) (report of four cases without ocular involve-
24. Esquivel CO, Mieles L, Marino IR, et al. Liver transplanta- ment). Br J Dermatol 1981;104:469475
tion for hereditary tyrosinemia in the presence of hepatocel- 45. Bienfang DC, Kuwabara T, Pueschel SM. The Richner-
lular carcinoma. Transplant Proc 1989;21:24452446 Hanhart syndrome: report of a case with associated tyrosine-
25. van Thiel DH, Gartner LM, Thorp FK, et al. Resolution of mia. Arch Ophthalmol 1976;94:11331137
the clinical features of tyrosinemia following orthotopic liver 46. Endo F, Katoh H, Matsuda I. Putative genetic deficiency of
transplantation for hepatoma. J Hepatol 1986;3:4248 4-hydroxyphenylpyruvic acid dioxygenase in mice: a murine
26. Horwich AL. Inherited hepatic enzyme defects as candidates model for hereditary tyrosinaemia type III. J Inherit Metab
for liver-directed gene therapy. Curr Top Microbiol Immunol Dis 1990;13:780782
1991;168:185200 47. Cerone R, Holme E, Schiaffino MC, et al. Tyrosinemia type
27. Lock EA, Ellis MK, Gaskin P, et al. From toxicological prob- III: diagnosis and ten-year follow-up. Acta Paediatr 1997;86:
lem to therapeutic use: the discovery of the mode of action of 10131015
2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione 48. Ruppert S, Kelsey G, Schedl A, et al. Deficiency of an en-
HEREDITARY TYROSINEMIA/GROMPE 571

zyme of tyrosine metabolism underlies altered gene expres- duction of delta-aminolevulinate synthase in liver. Trans
sion in newborn liver of lethal albino mice. Genes Dev Assoc Am Physicians 1982;95:4252
1992;6:14301443 66. Russell LB, Russell WL, Kelly EM. Analysis of the albino-
49. Kubo S, Sun M, Miyahara M, et al. Hepatocyte injury in ty- locus region of the mouse. Origin and viability. Genetics
rosinemia type 1 is induced by fumarylacetoacetate and is in- 1979;91:127139
hibited by caspase inhibitors. Proc Natl Acad Sci USA 1998; 67. Niswander L, Kelsey G, Schedl A, et al. Molecular mapping
95:95529557 of albino deletions associated with early embryonic lethality
50. Sun MS, Hattori S, Kubo S, et al. A mouse model of renal tu- in the mouse. Genomics 1991;9:162169

Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.
bular injury of tyrosinemia type 1: development of de Toni 68. Grompe M, Al-Dhalimy M, Finegold M, et al. Loss of fu-
Fanconi syndrome and apoptosis of renal tubular cells in marylacetoacetate hydrolase is responsible for the neonatal
Fah/Hpd double mutant mice. J Am Soc Nephrol 2000;11: hepatic dysfunction phenotype of lethal albino mice. Genes
291300 Dev 1993;7:22982307
51. Jorquera R, Tanguay RM. Cyclin B-dependent kinase and 69. Grompe M, Lindstedt S, al-Dhalimy M, et al. Pharmacologi-
caspase-1 activation precedes mitochondrial dysfunction in cal correction of neonatal lethal hepatic dysfunction in a
fumarylacetoacetate-induced apoptosis. FASEB J 1999;13: murine model of hereditary tyrosinaemia type I. Nat Genet
22842298 1995;10:453460
52. Schmid W, Mller G, Schtz G, et al. Deletions near the al- 70. Grompe M, Overturf K, Al-Dhalimy M, et al. Therapeutic
bino locus on chromosome 7 of the mouse affect the level of trials in the murine model of hereditary tyrosinemia type I: a
tyrosine aminotransferase mRNA. Proc Natl Acad Sci USA progress report. J Inher Metab Dis 1998;21:518531
1985;82:28662869 71. Overturf K, al-Dhalimy M, Ou CN, et al. Adenovirus-
53. Gluecksohn-Waelsch S. Genetic control of morphogenetic mediated gene therapy in a mouse model of hereditary ty-
and biochemical differentiation: lethal albino deletions in the rosinemia type I. Hum Gene Ther 1997;8:513521
mouse. Cell 1979;16:225237 72. Overturf K, Al-Dhalimy M, Manning K, et al. Ex vivo he-
54. Loose DS, Shaw PA, Krauter KS, et al. Trans regulation of patic gene therapy of a mouse model of hereditary tyrosine-
the phosphoenolpyruvate carboxykinase (GTP) gene, identi- mia type I. Hum Gene Ther 1998;9:295304
fied by deletions in chromosome 7 of the mouse. Proc Natl 73. Chen SJ, Tazelaar J, Moscioni AD, et al. In vivo selection of
Acad Sci USA 1986;83:51845188 hepatocytes transduced with adeno-associated viral vectors.
55. Sala-Trepat JM, Poiret M, Sellem CH, Bessada R, et al. A Mol Ther 2000;1:414422
lethal deletion on mouse chromosome 7 affects regulation of 74. Overturf K, Al-Dhalimy M, Finegold M, et al. The repop-
liver cell-specific functions: posttranscriptional control of ulation potential of hepatocyte populations differing in size
serum protein and transcriptional control of aldolase B syn- and prior mitotic expansion. Am J Pathol 1999;155:
thesis. Proc Natl Acad Sci USA 1985;82:24422446 21352143
56. Ruppert S, Boshart M, Bosch FX, et al. Two genetically de- 75. Grompe M, Al-Dhalimy M, Overturf K, et al. Hepatic re-
fined trans-acting loci coordinately regulate overlapping sets population by adult murine pancreatic liver stem cells. Am J
of liver-specific genes. Cell 1990;61:895904 Hum Genet 1998;63:9
57. Fornace AJ Jr, Nebert DW, Hollander MC, et al. Mammalian 76. Overturf K, Al-Dhalimy M, Ou CN, et al. Serial transplanta-
genes coordinately regulated by growth arrest signals and tion reveals the stem-celllike regenerative potential of adult
DNA-damaging agents. Mol Cell Biol 1989;9:4196203 mouse hepatocytes. Am J Pathol 1997;151:12731280
58. Petersen DD, Gonzalez FJ, Rapic V, et al. Marked increases 77. Wang X, Al-Dhalimy M, Lagasse E, et al. Liver repopulation
in hepatic NAD(P)H: oxidoreductase gene transcription and and correction of metabolic liver disease by transplanted adult
mRNA levels correlated with a mouse chromosome 7 dele- mouse pancreatic cells. Am J Pathol 2001;158:571579
tion. Proc Natl Acad Sci USA 1989;86:66996703 78. Lagasse E, Connors H, Al-Dhalimy M, et al. Purified
59. Nebert DW, Roe AL, Dieter MZ, et al. Role of the aromatic hematopoietic stem cells can differentiate into hepatocytes in
hydrocarbon receptor and [Ah] gene battery in the oxidative vivo. Nat Med 2000;6:12291234
stress response, cell cycle control, and apoptosis. Biochem 79. Grompe M. Liver repopulation for the treatment of meta-
Pharmacol 2000;59:6585 bolic diseases. J Inherit Metab Dis 2001;24:231244
60. Vasiliou V, Puga A, Chang CY, et al. Interaction between the 80. Grompe M, Laconi E, Shafritz DA. Principles of therapeutic
Ah receptor and proteins binding to the AP-1like electrophile liver repopulation. Semin Liver Dis 1999;19:714
response element (EpRE) during murine phase II [Ah] battery 81. Fernandez-Canon JM, Penalva MA. Fungal metabolic model
gene expression. Biochem Pharmacol 1995;50:20572068 for human type I hereditary tyrosinaemia. Proc Natl Acad Sci
61. Spencer PD, Roth KS. Effects of succinylacetone on amino USA 1995;92:91329136
acid uptake in the rat kidney. Biochem Med Metab Biol 82. Fernandez-Canon JM, Penalva MA. Molecular characteriza-
1987;37:101109 tion of a gene encoding a homogentisate dioxygenase from
62. Spencer PD, Medow MS, Moses LC, et al. Effects of suc- Aspergillus nidulans and identification of its human and plant
cinylacetone on the uptake of sugars and amino acids by homologues. J Biol Chem 1995;270:2119921205
brush border vesicles. Kidney Int 1988;34:671677 83. Fernandez-Canon JM, Penalva MA. Characterization of a
63. Jorquera R, Tanguay RM. The mutagenicity of the tyrosine fungal maleylacetoacetate isomerase gene and identification
metabolite, fumarylacetoacetate, is enhanced by glutathione of its human homologue. J Biol Chem 1998;273:329337
depletion. Biochem Biophys Res Commun 1997;232:4248 84. Garrod AE. The incidence of alkaptonuria: a study in chemi-
64. Manning K, Al-Dhalimy M, Finegold M, et al. In vivo suppres- cal individuality. Lancet 1902;2:16161620
sor mutations correct a murine model of hereditary tyrosinemia 85. Fernandez-Canon JM, Granadino B, Beltran-Valero de
type I. Proc Natl Acad Sci USA 1999;96:1192811933 Bernabe D, et al. The molecular basis of alkaptonuria. Nat
65. Sassa S, Kappas A. Succinylacetone inhibits delta-aminole- Genet 1996;14:1924
vulinate dehydratase and potentiates the drug and steroid in-
Downloaded by: IP-Proxy CONSORTIUM:Harrass (Oregon HealthScience Univ), Oregon Health and Science University. Copyrighted material.

Вам также может понравиться